146 results on '"Goyama S"'
Search Results
2. Protease-activated receptor-1 inhibits proliferation but enhances leukemia stem cell activity in acute myeloid leukemia
- Author
-
Goyama, S, Shrestha, M, Schibler, J, Rosenfeldt, L, Miller, W, O’Brien, E, Mizukawa, B, Kitamura, T, Palumbo, J S, and Mulloy, J C
- Published
- 2017
- Full Text
- View/download PDF
3. UBASH3B/Sts-1-CBL axis regulates myeloid proliferation in human preleukemia induced by AML1-ETO
- Author
-
Goyama, S, Schibler, J, Gasilina, A, Shrestha, M, Lin, S, Link, K A, Chen, J, Whitman, S P, Bloomfield, C D, Nicolet, D, Assi, S A, Ptasinska, A, Heidenreich, O, Bonifer, C, Kitamura, T, Nassar, N N, and Mulloy, J C
- Published
- 2016
- Full Text
- View/download PDF
4. Posttranslational modifications of RUNX1 as potential anticancer targets
- Author
-
Goyama, S, Huang, G, Kurokawa, M, and Mulloy, J C
- Published
- 2015
- Full Text
- View/download PDF
5. EVI-1 interacts with histone methyltransferases SUV39H1 and G9a for transcriptional repression and bone marrow immortalization
- Author
-
Goyama, S, Nitta, E, Yoshino, T, Kako, S, Watanabe-Okochi, N, Shimabe, M, Imai, Y, Takahashi, K, and Kurokawa, M
- Published
- 2010
- Full Text
- View/download PDF
6. Evi1 defines leukemia-initiating capacity and tyrosine kinase inhibitor resistance in chronic myeloid leukemia
- Author
-
Sato, T, Goyama, S, Kataoka, K, Nasu, R, Tsuruta-Kishino, T, Kagoya, Y, Nukina, A, Kumagai, K, Kubota, N, Nakagawa, M, Arai, S, Yoshimi, A, Honda, H, Kadowaki, T, and Kurokawa, M
- Published
- 2014
- Full Text
- View/download PDF
7. Pbx1 is a downstream target of Evi-1 in hematopoietic stem/progenitors and leukemic cells
- Author
-
Shimabe, M, Goyama, S, Watanabe-Okochi, N, Yoshimi, A, Ichikawa, M, Imai, Y, and Kurokawa, M
- Published
- 2009
- Full Text
- View/download PDF
8. AML1-Evi-1 specifically transforms hematopoietic stem cells through fusion of the entire Evi-1 sequence to AML1
- Author
-
Takeshita, M, Ichikawa, M, Nitta, E, Goyama, S, Asai, T, Ogawa, S, Chiba, S, and Kurokawa, M
- Published
- 2008
- Full Text
- View/download PDF
9. Male predominance among Japanese adult patients with late-onset hemorrhagic cystitis after hematopoietic stem cell transplantation
- Author
-
Asano, Y, Kanda, Y, Ogawa, N, Sakata-Yanagimoto, M, Nakagawa, M, Kawazu, M, Goyama, S, Kandabashi, K, Izutsu, K, Imai, Y, Hangaishi, A, Kurokawa, M, Tsujino, S, Ogawa, S, Aoki, K, Chiba, S, Motokura, T, and Hirai, H
- Published
- 2003
- Full Text
- View/download PDF
10. Post-transplant endothelial disorder after hematopoietic SCT: a blinded autopsy study
- Author
-
Goyama, S, Takeuchi, K, Kanda, Y, Nannya, Y, Chiba, S, Fukayama, M, and Kurokawa, M
- Published
- 2012
- Full Text
- View/download PDF
11. High expression of ABCG2 induced by EZH2 disruption has pivotal roles in MDS pathogenesis
- Author
-
Kawabata, K C, primary, Hayashi, Y, additional, Inoue, D, additional, Meguro, H, additional, Sakurai, H, additional, Fukuyama, T, additional, Tanaka, Y, additional, Asada, S, additional, Fukushima, T, additional, Nagase, R, additional, Takeda, R, additional, Harada, Y, additional, Kitaura, J, additional, Goyama, S, additional, Harada, H, additional, Aburatani, H, additional, and Kitamura, T, additional
- Published
- 2017
- Full Text
- View/download PDF
12. Protease-activated receptor-1 inhibits proliferation but enhances leukemia stem cell activity in acute myeloid leukemia
- Author
-
Goyama, S, primary, Shrestha, M, additional, Schibler, J, additional, Rosenfeldt, L, additional, Miller, W, additional, O’Brien, E, additional, Mizukawa, B, additional, Kitamura, T, additional, Palumbo, J S, additional, and Mulloy, J C, additional
- Published
- 2016
- Full Text
- View/download PDF
13. UBASH3B/Sts-1-CBL axis regulates myeloid proliferation in human preleukemia induced by AML1-ETO
- Author
-
Goyama, S, primary, Schibler, J, additional, Gasilina, A, additional, Shrestha, M, additional, Lin, S, additional, Link, K A, additional, Chen, J, additional, Whitman, S P, additional, Bloomfield, C D, additional, Nicolet, D, additional, Assi, S A, additional, Ptasinska, A, additional, Heidenreich, O, additional, Bonifer, C, additional, Kitamura, T, additional, Nassar, N N, additional, and Mulloy, J C, additional
- Published
- 2015
- Full Text
- View/download PDF
14. The subtype-specific features of EVI1 and PRDM16 in acute myeloid leukemia
- Author
-
Matsuo, H., primary, Goyama, S., additional, Kamikubo, Y., additional, and Adachi, S., additional
- Published
- 2015
- Full Text
- View/download PDF
15. Posttranslational modifications of RUNX1 as potential anticancer targets
- Author
-
Goyama, S, primary, Huang, G, additional, Kurokawa, M, additional, and Mulloy, J C, additional
- Published
- 2014
- Full Text
- View/download PDF
16. High expression of ABCG2 induced by EZH2 disruption has pivotal roles in MDS pathogenesis
- Author
-
Kawabata, K C, Hayashi, Y, Inoue, D, Meguro, H, Sakurai, H, Fukuyama, T, Tanaka, Y, Asada, S, Fukushima, T, Nagase, R, Takeda, R, Harada, Y, Kitaura, J, Goyama, S, Harada, H, Aburatani, H, and Kitamura, T
- Abstract
Both proto-oncogenic and tumor-suppressive functions have been reported for enhancer of zeste homolog 2 (EZH2). To investigate the effects of its inactivation, a mutant EZH2 lacking its catalytic domain was prepared (EZH2-dSET). In a mouse bone marrow transplant model, EZH2-dSET expression in bone marrow cells induced a myelodysplastic syndrome (MDS)-like disease in transplanted mice. Analysis of these mice identified Abcg2 as a direct target of EZH2. Intriguingly, Abcg2 expression alone induced the same disease in the transplanted mice, where stemness genes were enriched. Interestingly, ABCG2 expression is specifically high in MDS patients. The present results indicate that ABCG2 de-repression induced by EZH2 mutations have crucial roles in MDS pathogenesis.
- Published
- 2018
- Full Text
- View/download PDF
17. Post-transplant endothelial disorder after hematopoietic SCT: a blinded autopsy study
- Author
-
Goyama, S, primary, Takeuchi, K, additional, Kanda, Y, additional, Nannya, Y, additional, Chiba, S, additional, Fukayama, M, additional, and Kurokawa, M, additional
- Published
- 2011
- Full Text
- View/download PDF
18. EVI-1 interacts with histone methyltransferases SUV39H1 and G9a for transcriptional repression and bone marrow immortalization
- Author
-
Goyama, S, primary, Nitta, E, additional, Yoshino, T, additional, Kako, S, additional, Watanabe-Okochi, N, additional, Shimabe, M, additional, Imai, Y, additional, Takahashi, K, additional, and Kurokawa, M, additional
- Published
- 2009
- Full Text
- View/download PDF
19. Reverse Seroconversion of Hepatitis B Virus after Hematopoietic Stem Cell Transplantation
- Author
-
Goyama, S., primary, Kanda, Y., additional, Nannya, Y., additional, Kawazu, M., additional, Takeshita, M., additional, Niino, M., additional, Komeno, Y., additional, Nakamoto, T., additional, Kurokawa, M., additional, Tsujino, S., additional, Ogawa, S., additional, Aoki, K., additional, Chiba, S., additional, Motokura, T., additional, Shiratori, Y., additional, and Hirai, H., additional
- Published
- 2002
- Full Text
- View/download PDF
20. Nucleic Acid Metabolism: the Key Therapeutic Target for Myeloid Tumors.
- Author
-
Yabushita T and Goyama S
- Abstract
Nucleic acid analogs, including cytarabine, decitabine, and azacitidine, have significantly advanced therapeutic approaches for myeloid tumors over the past five decades. Nucleic acid metabolism is a crucial pathway driving myeloid tumorigenesis, with emerging evidence indicating that myeloid tumors are particularly dependent on the de novo nucleotide synthesis pathway, underscoring its potential as a therapeutic target. This review provides a comprehensive overview of nucleic acid metabolism, with a particular focus on de novo nucleotide synthesis. We then describe the range of clinically utilized agents targeting nucleic acid metabolism and discuss our recent findings on the non-epigenetic actions of decitabine, as well as the therapeutic effects of IMPDH inhibitors in the treatment of myeloid tumors., Competing Interests: Declaration of competing interest The authors declare they have no conflicts of interest with respect to this article., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
21. Clonal hematopoiesis-related mutant ASXL1 promotes atherosclerosis in mice via dysregulated innate immunity.
- Author
-
Sato N, Goyama S, Chang YH, Miyawaki M, Fujino T, Koide S, Denda T, Liu X, Ueda K, Yamamoto K, Asada S, Takeda R, Yonezawa T, Tanaka Y, Honda H, Ota Y, Shibata T, Sekiya M, Isobe T, Lamagna C, Masuda E, Iwama A, Shimano H, Inoue JI, Miyake K, and Kitamura T
- Subjects
- Animals, MAP Kinase Kinase Kinases genetics, MAP Kinase Kinase Kinases metabolism, Mice, Knockout, Mice, Mice, Inbred C57BL, Monocytes immunology, Monocytes metabolism, NF-kappa B metabolism, NF-kappa B genetics, Macrophages immunology, Macrophages metabolism, Humans, Male, Plaque, Atherosclerotic genetics, Plaque, Atherosclerotic pathology, Plaque, Atherosclerotic immunology, Aortic Diseases genetics, Aortic Diseases immunology, Aortic Diseases pathology, Atherosclerosis genetics, Atherosclerosis immunology, Atherosclerosis pathology, Atherosclerosis metabolism, Immunity, Innate genetics, Repressor Proteins genetics, Repressor Proteins metabolism, Interleukin-1 Receptor-Associated Kinases genetics, Interleukin-1 Receptor-Associated Kinases metabolism, Mutation, Disease Models, Animal, Signal Transduction genetics, Clonal Hematopoiesis genetics
- Abstract
Certain somatic mutations provide a fitness advantage to hematopoietic stem cells and lead to clonal expansion of mutant blood cells, known as clonal hematopoiesis (CH). Among the most common CH mutations, ASXL1 mutations pose the highest risk for cardiovascular diseases (CVDs), yet the mechanisms by which they contribute to CVDs are unclear. Here we show that hematopoietic cells harboring C-terminally truncated ASXL1 mutant (ASXL1-MT) accelerate the development of atherosclerosis in Ldlr
-/- mice. Transcriptome analyses of plaque cells showed that monocytes and macrophages expressing ASXL1-MT exhibit inflammatory signatures. Mechanistically, we demonstrate that wild-type ASXL1 has an unexpected non-epigenetic role by suppressing innate immune signaling through the inhibition of IRAK1-TAK1 interaction in the cytoplasm. This regulatory function is lost in ASXL1-MT, resulting in NF-κB activation. Inhibition of IRAK1/4 alleviated atherosclerosis driven by ASXL1-MT and decreased inflammatory monocytes. The present work provides a mechanistic and cellular explanation linking ASXL1 mutations, CH and CVDs., Competing Interests: Competing interests: E.M. and C.L. own stock in and are employees of Rigel Pharmaceuticals, Inc. The other authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
22. HDAC7 is a potential therapeutic target in acute erythroid leukemia.
- Author
-
Zhang W, Yamamoto K, Chang YH, Yabushita T, Hao Y, Shimura R, Nakahara J, Shikata S, Iida K, Chen Q, Zhang X, Kitamura T, and Goyama S
- Subjects
- Animals, Mice, Humans, Cell Proliferation, Transcriptional Regulator ERG genetics, Transcriptional Regulator ERG metabolism, CRISPR-Cas Systems, Disease Models, Animal, Leukemia, Erythroblastic, Acute pathology, Leukemia, Erythroblastic, Acute genetics, Leukemia, Erythroblastic, Acute metabolism, Histone Deacetylases metabolism, Histone Deacetylases genetics, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Protein p53 genetics
- Abstract
Acute erythroleukemia (AEL) is a rare subtype of acute myeloid leukemia with a poor prognosis. In this study, we established a novel murine AEL model with Trp53 depletion and ERG overexpression. ERG overexpression in Trp53-deficient mouse bone marrow cells, but not in wild-type bone marrow cells, leads to AEL development within two months after transplantation with 100% penetrance. The established mouse AEL cells expressing Cas9 can be cultured in vitro, induce AEL in vivo even in unirradiated recipient mice, and enable efficient gene ablation using the CRISPR/Cas9 system. We also confirmed the cooperation between ERG overexpression and TP53 inactivation in promoting the growth of immature erythroid cells in human cord blood cells. Mechanistically, ERG antagonizes KLF1 and inhibits erythroid maturation, whereas TP53 deficiency promotes proliferation of erythroid progenitors. Furthermore, we identified HDAC7 as a specific susceptibility in AEL by the DepMap-based two-group comparison analysis. HDAC7 promotes the growth of human and mouse AEL cells both in vitro and in vivo through its non-enzymatic functions. Our study provides experimental evidence that TP53 deficiency and ERG overexpression are necessary and sufficient for the development of AEL and highlights HDAC7 as a promising therapeutic target for this disease., Competing Interests: Competing interests: The authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
23. NPM1-fusion proteins promote myeloid leukemogenesis through XPO1-dependent HOX activation.
- Author
-
Shimosato Y, Yamamoto K, Jia Y, Zhang W, Shiba N, Hayashi Y, Ito S, Kitamura T, and Goyama S
- Abstract
Nucleophosmin (NPM1) is a nucleolar protein and one of the most frequently mutated genes in acute myeloid leukemia (AML). In addition to the commonly detected frameshift mutations in exon12 (NPM1c), previous studies have identified NPM1 gene rearrangements leading to the expression of NPM1-fusion proteins in pediatric AML. However, whether the NPM1-fusions are indeed oncogenic and how the NPM1-fusions cause AML have been largely unknown. In this study, we investigated the subcellular localization and leukemogenic potential of two rare NPM1-fusion proteins, NPM1::MLF1 and NPM1::CCDC28A. NPM1::MLF1 is present in both the nucleus and cytoplasm and occasionally induces AML in the mouse transplantation assay. NPM1::CCDC28A is more localized to the cytoplasm, immortalizes mouse bone marrow cells in vitro and efficiently induces AML in vivo. Mechanistically, both NPM1-fusions bind to the HOX gene cluster and, like NPM1c, cause aberrant upregulation of HOX genes in cooperation with XPO1. The XPO1 inhibitor selinexor suppressed HOX activation and colony formation driven by the NPM1-fusions. NPM1::CCDC28A cells were also sensitive to menin inhibition. Thus, our study provides experimental evidence that both NPM1::MLF1 and NPM1::CCDC28A are oncogenes with functions similar to NPM1c. Inhibition of XPO1 and menin may be a promising strategy for the NPM1-rearranged AML., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
24. Isolation of Mitotic Centrosomes from Cultured Human Cells.
- Author
-
Miyazawa M, Yamamoto S, Goyama S, and Kitagawa D
- Subjects
- Humans, Microtubules metabolism, Spindle Apparatus metabolism, Cell Fractionation methods, Cells, Cultured, Centrosome metabolism, Mitosis
- Abstract
The centrosome plays a crucial role in facilitating mitotic spindle assembly through its microtubule organizing capacities. Analyzing the composition, structure, and functions of mitotic centrosomes is essential for understanding the mechanisms underlying cell division and centrosome-associated diseases. Isolating centrosomes is an effective method to gain comprehensive information about them while minimizing interference from other cellular components. In this chapter, we describe a protocol for isolating mitotic centrosomes from cultured human cells. This protocol includes cell synchronization and centrosome isolation through ultracentrifugation with a sucrose gradient. We also describe a method for conducting a microtubule nucleation assay to assess the functionality of isolated centrosomes., (© 2025. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2025
- Full Text
- View/download PDF
25. Inhibition of TOPORS ubiquitin ligase augments the efficacy of DNA hypomethylating agents through DNMT1 stabilization.
- Author
-
Kaito S, Aoyama K, Oshima M, Tsuchiya A, Miyota M, Yamashita M, Koide S, Nakajima-Takagi Y, Kozuka-Hata H, Oyama M, Yogo T, Yabushita T, Ito R, Ueno M, Hirao A, Tohyama K, Li C, Kawabata KC, Yamaguchi K, Furukawa Y, Kosako H, Yoshimi A, Goyama S, Nannya Y, Ogawa S, Agger K, Helin K, Yamazaki S, Koseki H, Doki N, Harada Y, Harada H, Nishiyama A, Nakanishi M, and Iwama A
- Subjects
- Humans, Animals, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases genetics, Cell Line, Tumor, Mice, CRISPR-Cas Systems, HEK293 Cells, DNA (Cytosine-5-)-Methyltransferase 1 metabolism, DNA (Cytosine-5-)-Methyltransferase 1 antagonists & inhibitors, DNA (Cytosine-5-)-Methyltransferase 1 genetics, Ubiquitination drug effects, Sumoylation drug effects, DNA Methylation drug effects
- Abstract
DNA hypomethylating agents (HMAs) are used for the treatment of myeloid malignancies, although their therapeutic effects have been unsatisfactory. Here we show that CRISPR-Cas9 screening reveals that knockout of topoisomerase 1-binding arginine/serine-rich protein (TOPORS), which encodes a ubiquitin/SUMO E3 ligase, augments the efficacy of HMAs on myeloid leukemic cells with little effect on normal hematopoiesis, suggesting that TOPORS is involved in resistance to HMAs. HMAs are incorporated into the DNA and trap DNA methyltransferase-1 (DNMT1) to form DNA-DNMT1 crosslinks, which undergo SUMOylation, followed by proteasomal degradation. Persistent crosslinking is cytotoxic. The TOPORS RING finger domain, which mediates ubiquitination, is responsible for HMA resistance. In TOPORS knockout cells, DNMT1 is stabilized by HMA treatment due to inefficient ubiquitination, resulting in the accumulation of unresolved SUMOylated DNMT1. This indicates that TOPORS ubiquitinates SUMOylated DNMT1, thereby promoting the resolution of DNA-DNMT1 crosslinks. Consistently, the ubiquitination inhibitor, TAK-243, and the SUMOylation inhibitor, TAK-981, show synergistic effects with HMAs through DNMT1 stabilization. Our study provides a novel HMA-based therapeutic strategy that interferes with the resolution of DNA-DNMT1 crosslinks., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
26. SETDB1 suppresses NK cell-mediated immunosurveillance in acute myeloid leukemia with granulo-monocytic differentiation.
- Author
-
Chang YH, Yamamoto K, Fujino T, Wang TW, Sugimoto E, Zhang W, Yabushita T, Suzaki K, Pietsch EC, Weir BA, Crescenzo R, Cowley GS, Attar R, Philippar U, Wunderlich M, Mizukawa B, Zheng Y, Enomoto Y, Imai Y, Kitamura T, and Goyama S
- Subjects
- Animals, Mice, Humans, Mice, Inbred C57BL, Cell Line, Tumor, Immunologic Surveillance, Monocytes metabolism, Monocytes immunology, Apoptosis, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute genetics, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Cell Differentiation, Histone-Lysine N-Methyltransferase metabolism, Histone-Lysine N-Methyltransferase genetics
- Abstract
Monocytic acute myeloid leukemia (AML) responds poorly to current treatments, including venetoclax-based therapy. We conducted in vivo and in vitro CRISPR-Cas9 library screenings using a mouse monocytic AML model and identified SETDB1 and its binding partners (ATF7IP and TRIM33) as crucial tumor promoters in vivo. The growth-inhibitory effect of Setdb1 depletion in vivo is dependent mainly on natural killer (NK) cell-mediated cytotoxicity. Mechanistically, SETDB1 depletion upregulates interferon-stimulated genes and NKG2D ligands through the demethylation of histone H3 Lys9 at the enhancer regions, thereby enhancing their immunogenicity to NK cells and intrinsic apoptosis. Importantly, these effects are not observed in non-monocytic leukemia cells. We also identified the expression of myeloid cell nuclear differentiation antigen (MNDA) and its murine counterpart Ifi203 as biomarkers to predict the sensitivity of AML to SETDB1 depletion. Our study highlights the critical and selective role of SETDB1 in AML with granulo-monocytic differentiation and underscores its potential as a therapeutic target for current unmet needs., Competing Interests: Declaration of interests T.K. has received research support from Janssen Research and Development for this study. E.C.P., R.C., G.S.C., B.A.W., R.A., and U.P. are employees of Janssen Research and Development., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
27. Modeling and therapeutic targeting of t(8;21) AML with/without TP53 deficiency.
- Author
-
Zhang W, Li J, Yamamoto K, and Goyama S
- Subjects
- Animals, Mice, Humans, Oncogene Proteins, Fusion genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute etiology, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 deficiency, Chromosomes, Human, Pair 8 genetics, Translocation, Genetic, Chromosomes, Human, Pair 21 genetics, Disease Models, Animal, Core Binding Factor Alpha 2 Subunit genetics, Core Binding Factor Alpha 2 Subunit deficiency
- Abstract
Acute myeloid leukemia (AML) with t(8;21)(q22;q22.1);RUNX1-ETO is one of the most common subtypes of AML. Although t(8;21) AML has been classified as favorable-risk, only about half of patients are cured with current therapies. Several genetic abnormalities, including TP53 mutations and deletions, negatively impact survival in t(8;21) AML. In this study, we established Cas9
+ mouse models of t(8;21) AML with intact or deficient Tpr53 (a mouse homolog of TP53) using a retrovirus-mediated gene transfer and transplantation system. Trp53 deficiency accelerates the in vivo development of AML driven by RUNX1-ETO9a, a short isoform of RUNX1-ETO with strong leukemogenic potential. Trp53 deficiency also confers resistance to genetic depletion of RUNX1 and a TP53-activating drug in t(8;21) AML. However, Trp53-deficient t(8;21) AML cells were still sensitive to several drugs such as dexamethasone. Cas9+ RUNX1-ETO9a cells with/without Trp53 deficiency can produce AML in vivo, can be cultured in vitro for several weeks, and allow efficient gene depletion using the CRISPR/Cas9 system, providing useful tools to advance our understanding of t(8;21) AML., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
28. Integrative analysis of cancer multimodality data identifying COPS5 as a novel biomarker of diffuse large B-cell lymphoma.
- Author
-
Dai Y, Li J, Yamamoto K, Goyama S, Loza M, Park SJ, and Nakai K
- Abstract
Preventing, diagnosing, and treating diseases requires accurate clinical biomarkers, which remains challenging. Recently, advanced computational approaches have accelerated the discovery of promising biomarkers from high-dimensional multimodal data. Although machine-learning methods have greatly contributed to the research fields, handling data sparseness, which is not unusual in research settings, is still an issue as it leads to limited interpretability and performance in the presence of missing information. Here, we propose a novel pipeline integrating joint non-negative matrix factorization (JNMF), identifying key features within sparse high-dimensional heterogeneous data, and a biological pathway analysis, interpreting the functionality of features by detecting activated signaling pathways. By applying our pipeline to large-scale public cancer datasets, we identified sets of genomic features relevant to specific cancer types as common pattern modules (CPMs) of JNMF. We further detected COPS5 as a potential upstream regulator of pathways associated with diffuse large B-cell lymphoma (DLBCL). COPS5 exhibited co-overexpression with MYC , TP53 , and BCL2 , known DLBCL marker genes, and its high expression was correlated with a lower survival probability of DLBCL patients. Using the CRISPR-Cas9 system, we confirmed the tumor growth effect of COPS5 , which suggests it as a novel prognostic biomarker for DLBCL. Our results highlight that integrating multiple high-dimensional data and effectively decomposing them to interpretable dimensions unravels hidden biological importance, which enhances the discovery of clinical biomarkers., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Dai, Li, Yamamoto, Goyama, Loza, Park and Nakai.)
- Published
- 2024
- Full Text
- View/download PDF
29. Immune checkpoint molecule DNAM-1/CD112 axis is a novel target for natural killer-cell therapy in acute myeloid leukemia.
- Author
-
Kaito Y, Sugimoto E, Nakamura F, Tsukune Y, Sasaki M, Yui S, Yamaguchi H, Goyama S, Nannya Y, Mitani K, Tamura H, and Imai Y
- Subjects
- Humans, Nectins, Killer Cells, Natural, Receptors, Immunologic, Cell- and Tissue-Based Therapy, Antigens, Differentiation, T-Lymphocyte genetics, Antigens, Differentiation, T-Lymphocyte metabolism, Immune Checkpoint Proteins metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute metabolism
- Abstract
Acute myeloid leukemia (AML) is a hematologic malignancy that frequently relapses, even if remission can be achieved with intensive chemotherapy. One known relapse mechanism is the escape of leukemic cells from immune surveillance. Currently, there is no effective immunotherapy for AML because of the lack of specific antigens. Here, we aimed to elucidate the association between CD155 and CD112 in AML cell lines and primary AML samples and determine the therapeutic response. Briefly, we generated NK-92 cell lines (NK-92) with modified DNAX-associated molecule 1 (DNAM-1) and T-cell immunoglobulin and ITIM domain (TIGIT), which are receptors of CD155 and CD112, respectively. Analysis of 200 cases of AML indicated that the survival of patients with high expression of CD112 was shorter than that of patients with low expression. NK-92 DNAM-1 exhibited enhanced cytotoxic activity against AML cell lines and primary cells derived from patients with AML. DNAM-1 induction in NK-92 cells enhanced the expression of cytotoxicity-related genes, thus overcoming the inhibitory activity of TIGIT. Between CD155 and CD112, CD112 is an especially important target for natural killer (NK)-cell therapy of AML. Using a xenograft model, we confirmed the enhanced antitumor effect of NK-92 DNAM-1 compared with that of NK-92 alone. We also discovered that CD112 (Nectin-2), an immune checkpoint molecule belonging to the Nectin/Nectin-like family, functions as a novel target of immunotherapy. In conclusion, modification of the DNAM-1/CD112 axis in NK cells may be an effective novel immunotherapy for AML. Furthermore, our findings suggest that the levels of expression of these molecules are potential prognostic markers in AML.
- Published
- 2024
- Full Text
- View/download PDF
30. [Molecular mechanism of DNA hypomethylating agents in myeloid tumors].
- Author
-
Yabushita T, Kitamura T, and Goyama S
- Subjects
- Humans, Azacitidine therapeutic use, Azacitidine pharmacology, Decitabine pharmacology, Decitabine therapeutic use, Animals, Molecular Targeted Therapy, DNA Methylation
- Published
- 2024
- Full Text
- View/download PDF
31. Hyperactive Natural Killer cells in Rag2 knockout mice inhibit the development of acute myeloid leukemia.
- Author
-
Sugimoto E, Li J, Hayashi Y, Iida K, Asada S, Fukushima T, Tamura M, Shikata S, Zhang W, Yamamoto K, Kawabata KC, Kawase T, Saito T, Yoshida T, Yamazaki S, Kaito Y, Imai Y, Denda T, Ota Y, Fukuyama T, Tanaka Y, Enomoto Y, Kitamura T, and Goyama S
- Subjects
- Animals, Mice, Mice, Knockout, Mice, Inbred C57BL, T-Lymphocytes, DNA-Binding Proteins genetics, Killer Cells, Natural, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology
- Abstract
Immunotherapy has attracted considerable attention as a therapeutic strategy for cancers including acute myeloid leukemia (AML). In this study, we found that the development of several aggressive subtypes of AML is slower in Rag2
-/- mice despite the lack of B and T lymphocytes, even compared to the immunologically normal C57BL/6 mice. Furthermore, an orally active p53-activating drug shows stronger antileukemia effect on AML in Rag2-/- mice than C57BL/6 mice. Intriguingly, Natural Killer (NK) cells in Rag2-/- mice are increased in number, highly express activation markers, and show increased cytotoxicity to leukemia cells in a coculture assay. B2m depletion that triggers missing-self recognition of NK cells impairs the growth of AML cells in vivo. In contrast, NK cell depletion accelerates AML progression in Rag2-/- mice. Interestingly, immunogenicity of AML keeps changing during tumor evolution, showing a trend that the aggressive AMLs generate through serial transplantations are susceptible to NK cell-mediated tumor suppression in Rag2-/- mice. Thus, we show the critical role of NK cells in suppressing the development of certain subtypes of AML using Rag2-/- mice, which lack functional lymphocytes but have hyperactive NK cells., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
32. BRD9 determines the cell fate of hematopoietic stem cells by regulating chromatin state.
- Author
-
Xiao M, Kondo S, Nomura M, Kato S, Nishimura K, Zang W, Zhang Y, Akashi T, Viny A, Shigehiro T, Ikawa T, Yamazaki H, Fukumoto M, Tanaka A, Hayashi Y, Koike Y, Aoyama Y, Ito H, Nishikawa H, Kitamura T, Kanai A, Yokoyama A, Fujiwara T, Goyama S, Noguchi H, Lee SC, Toyoda A, Hinohara K, Abdel-Wahab O, and Inoue D
- Subjects
- Chromatin Assembly and Disassembly, Cell Differentiation, Hematopoietic Stem Cells metabolism, Transcription Factors genetics, Transcription Factors metabolism, Chromatin
- Abstract
ATP-dependent chromatin remodeling SWI/SNF complexes exist in three subcomplexes: canonical BAF (cBAF), polybromo BAF (PBAF), and a newly described non-canonical BAF (ncBAF). While cBAF and PBAF regulate fates of multiple cell types, roles for ncBAF in hematopoietic stem cells (HSCs) have not been investigated. Motivated by recent discovery of disrupted expression of BRD9, an essential component of ncBAF, in multiple cancers, including clonal hematopoietic disorders, we evaluate here the role of BRD9 in normal and malignant HSCs. BRD9 loss enhances chromatin accessibility, promoting myeloid lineage skewing while impairing B cell development. BRD9 significantly colocalizes with CTCF, whose chromatin recruitment is augmented by BRD9 loss, leading to altered chromatin state and expression of myeloid-related genes within intact topologically associating domains. These data uncover ncBAF as critical for cell fate specification in HSCs via three-dimensional regulation of gene expression and illuminate roles for ncBAF in normal and malignant hematopoiesis., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
33. Inhibition of PLK4 remodels histone methylation and activates the immune response via the cGAS-STING pathway in TP53-mutated AML.
- Author
-
Man CH, Lam W, Dang CC, Zeng XY, Zheng LC, Chan NN, Ng KL, Chan KC, Kwok TH, Ng TC, Leung WY, Huen MS, Wong CC, So CWE, Dou Z, Goyama S, Bray MR, Mak TW, and Leung AY
- Subjects
- Animals, Mice, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Mutation, Methylation, Nucleotidyltransferases metabolism, Immunity, Polyploidy, Histones metabolism, Leukemia, Myeloid, Acute pathology
- Abstract
Acute myeloid leukemia (AML) with TP53 mutation is one of the most lethal cancers and portends an extremely poor prognosis. Based on in silico analyses of druggable genes and differential gene expression in TP53-mutated AML, we identified pololike kinase 4 (PLK4) as a novel therapeutic target and examined its expression, regulation, pathogenetic mechanisms, and therapeutic potential in TP53-mutated AML. PLK4 expression was suppressed by activated p53 signaling in TP53 wild-type AML and was increased in TP53-mutated AML cell lines and primary samples. Short-term PLK4 inhibition induced DNA damage and apoptosis in TP53 wild-type AML. Prolonged PLK4 inhibition suppressed the growth of TP53-mutated AML and was associated with DNA damage, apoptosis, senescence, polyploidy, and defective cytokinesis. A hitherto undescribed PLK4/PRMT5/EZH2/H3K27me3 axis was demonstrated in both TP53 wild-type and mutated AML, resulting in histone modification through PLK4-induced PRMT5 phosphorylation. In TP53-mutated AML, combined effects of histone modification and polyploidy activated the cGAS-STING pathway, leading to secretion of cytokines and chemokines and activation of macrophages and T cells upon coculture with AML cells. In vivo, PLK4 inhibition also induced cytokine and chemokine expression in mouse recipients, and its combination with anti-CD47 antibody, which inhibited the "don't-eat-me" signal in macrophages, synergistically reduced leukemic burden and prolonged animal survival. The study shed important light on the pathogenetic role of PLK4 and might lead to novel therapeutic strategies in TP53-mutated AML., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
34. The E3 ligase DTX2 inhibits RUNX1 function by binding its C terminus and prevents the growth of RUNX1-dependent leukemia cells.
- Author
-
Yonezawa T, Takahashi H, Hao Y, Furukawa C, Tsuchiya A, Zhang W, Fukushima T, Fukuyama T, Sawasaki T, Kitamura T, and Goyama S
- Subjects
- Humans, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Gene Expression Regulation, Core Binding Factor Alpha 2 Subunit genetics, Core Binding Factor Alpha 2 Subunit metabolism, Leukemia genetics
- Abstract
Transcription factor RUNX1 plays important roles in hematopoiesis and leukemogenesis. RUNX1 function is tightly controlled through posttranslational modifications, including ubiquitination and acetylation. However, its regulation via ubiquitination, especially proteasome-independent ubiquitination, is poorly understood. We previously identified DTX2 as a RUNX1-interacting E3 ligase using a cell-free AlphaScreen assay. In this study, we examined whether DTX2 is involved in the regulation of RUNX1 using in vitro and ex vivo analyses. DTX2 bound to RUNX1 and other RUNX family members RUNX2 and RUNX3 through their C-terminal region. DTX2-induced RUNX1 ubiquitination did not result in RUNX1 protein degradation. Instead, we found that the acetylation of RUNX1, which is known to enhance the transcriptional activity of RUNX1, was inhibited in the presence of DTX2. Concomitantly, DTX2 reduced the RUNX1-induced activation of an MCSFR luciferase reporter. We also found that DTX2 induced RUNX1 cytoplasmic mislocalization. Moreover, DTX2 overexpression showed a substantial growth-inhibitory effect in RUNX1-dependent leukemia cell lines. Thus, our findings indicate a novel aspect of the ubiquitination and acetylation of RUNX1 that is modulated by DTX2 in a proteosome-independent manner., (© 2023 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2023
- Full Text
- View/download PDF
35. Identification of compounds that preferentially suppress the growth of T-cell acute lymphoblastic leukemia-derived cells.
- Author
-
Miyashita K, Yagi T, Kagaya N, Takechi A, Nakata C, Kanda R, Nuriya H, Tanegashima K, Hoyano S, Seki F, Yoshida C, Hachiro Y, Higashi T, Kitada N, Toya T, Kobayashi T, Najima Y, Goyama S, Maki SA, Kitamura T, Doki N, Shin-Ya K, and Hara T
- Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is one of the most frequently occurring cancers in children and is associated with a poor prognosis. Here, we performed large-scale screening of natural compound libraries to identify potential drugs against T-ALL. We identified three low-molecular-weight compounds (auxarconjugatin-B, rumbrin, and lavendamycin) that inhibited the proliferation of the T-ALL cell line CCRF-CEM, but not that of the B lymphoma cell line Raji in a low concentration range. Among them, auxarconjugatin-B and rumbrin commonly contained a polyenyl 3-chloropyrrol in their chemical structure, therefore we chose auxarconjugatin-B for further analyses. Auxarconjugatin-B suppressed the in vitro growth of five human T-ALL cell lines and two T-ALL patient-derived cells, but not that of adult T-cell leukemia patient-derived cells. Cultured normal T cells were several-fold resistant to auxarconjugatin-B. Auxarconjugatin-B and its synthetic analogue Ra#37 depolarized the mitochondrial membrane potential of CCRF-CEM cells within 3 h of treatment. These compounds are promising seeds for developing novel anti-T-ALL drugs., (© 2023 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.)
- Published
- 2023
- Full Text
- View/download PDF
36. Mitotic perturbation is a key mechanism of action of decitabine in myeloid tumor treatment.
- Author
-
Yabushita T, Chinen T, Nishiyama A, Asada S, Shimura R, Isobe T, Yamamoto K, Sato N, Enomoto Y, Tanaka Y, Fukuyama T, Satoh H, Kato K, Saitoh K, Ishikawa T, Soga T, Nannya Y, Fukagawa T, Nakanishi M, Kitagawa D, Kitamura T, and Goyama S
- Subjects
- Humans, Decitabine pharmacology, Decitabine therapeutic use, Antimetabolites, Antineoplastic pharmacology, DNA Methylation genetics, DNA, Adaptor Proteins, Signal Transducing genetics, Azacitidine pharmacology, Azacitidine therapeutic use, Leukemia, Myeloid, Acute pathology
- Abstract
Decitabine (DAC) is clinically used to treat myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Our genome-wide CRISPR-dCas9 activation screen using MDS-derived AML cells indicates that mitotic regulation is critical for DAC resistance. DAC strongly induces abnormal mitosis (abscission failure or tripolar mitosis) in human myeloid tumors at clinical concentrations, especially in those with TP53 mutations or antecedent hematological disorders. This DAC-induced mitotic disruption and apoptosis are significantly attenuated in DNMT1-depleted cells. In contrast, overexpression of Dnmt1, but not the catalytically inactive mutant, enhances DAC-induced mitotic defects in myeloid tumors. We also demonstrate that DAC-induced mitotic disruption is enhanced by pharmacological inhibition of the ATR-CLSPN-CHK1 pathway. These data challenge the current assumption that DAC inhibits leukemogenesis through DNMT1 inhibition and subsequent DNA hypomethylation and highlight the potent activity of DAC to disrupt mitosis through aberrant DNMT1-DNA covalent bonds., Competing Interests: Declaration of interests The authors declare no potential conflicts of interest., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
37. TLR7/8 stress response drives histiocytosis in SLC29A3 disorders.
- Author
-
Shibata T, Sato R, Taoka M, Saitoh SI, Komine M, Yamaguchi K, Goyama S, Motoi Y, Kitaura J, Izawa K, Yamauchi Y, Tsukamoto Y, Ichinohe T, Fujita E, Hiranuma R, Fukui R, Furukawa Y, Kitamura T, Takai T, Tojo A, Ohtsuki M, Ohto U, Shimizu T, Ozawa M, Yoshida N, Isobe T, Latz E, Mukai K, Taguchi T, Hemmi H, Akira S, and Miyake K
- Subjects
- Animals, Mice, Cytokines genetics, Mutation genetics, Nucleosides, Toll-Like Receptor 8 genetics, Histiocytosis genetics, Toll-Like Receptor 7 genetics
- Abstract
Loss-of-function mutations in the lysosomal nucleoside transporter SLC29A3 cause lysosomal nucleoside storage and histiocytosis: phagocyte accumulation in multiple organs. However, little is known about the mechanism by which lysosomal nucleoside storage drives histiocytosis. Herein, histiocytosis in Slc29a3-/- mice was shown to depend on Toll-like receptor 7 (TLR7), which senses a combination of nucleosides and oligoribonucleotides (ORNs). TLR7 increased phagocyte numbers by driving the proliferation of Ly6Chi immature monocytes and their maturation into Ly6Clow phagocytes in Slc29a3-/- mice. Downstream of TLR7, FcRγ and DAP10 were required for monocyte proliferation. Histiocytosis is accompanied by inflammation in SLC29A3 disorders. However, TLR7 in nucleoside-laden splenic monocytes failed to activate inflammatory responses. Enhanced production of proinflammatory cytokines was observed only after stimulation with ssRNAs, which would increase lysosomal ORNs. Patient-derived monocytes harboring the G208R SLC29A3 mutation showed enhanced survival and proliferation in a TLR8-antagonist-sensitive manner. These results demonstrated that TLR7/8 responses to lysosomal nucleoside stress drive SLC29A3 disorders., (© 2023 Shibata et al.)
- Published
- 2023
- Full Text
- View/download PDF
38. IMPDH inhibition activates TLR-VCAM1 pathway and suppresses the development of MLL-fusion leukemia.
- Author
-
Liu X, Sato N, Yabushita T, Li J, Jia Y, Tamura M, Asada S, Fujino T, Fukushima T, Yonezawa T, Tanaka Y, Fukuyama T, Tsuchiya A, Shikata S, Iwamura H, Kinouchi C, Komatsu K, Yamasaki S, Shibata T, Sasaki AT, Schibler J, Wunderlich M, O'Brien E, Mizukawa B, Mulloy JC, Sugiura Y, Takizawa H, Shibata T, Miyake K, Kitamura T, and Goyama S
- Subjects
- Humans, Enzyme Inhibitors pharmacology, NF-kappa B, Immunosuppressive Agents therapeutic use, Myeloid-Lymphoid Leukemia Protein metabolism, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology
- Abstract
Inosine monophosphate dehydrogenase (IMPDH) is a rate-limiting enzyme in de novo guanine nucleotide synthesis pathway. Although IMPDH inhibitors are widely used as effective immunosuppressants, their antitumor effects have not been proven in the clinical setting. Here, we found that acute myeloid leukemias (AMLs) with MLL-fusions are susceptible to IMPDH inhibitors in vitro. We also showed that alternate-day administration of IMPDH inhibitors suppressed the development of MLL-AF9-driven AML in vivo without having a devastating effect on immune function. Mechanistically, IMPDH inhibition induced overactivation of Toll-like receptor (TLR)-TRAF6-NF-κB signaling and upregulation of an adhesion molecule VCAM1, which contribute to the antileukemia effect of IMPDH inhibitors. Consequently, combined treatment with IMPDH inhibitors and the TLR1/2 agonist effectively inhibited the development of MLL-fusion AML. These findings provide a rational basis for clinical testing of IMPDH inhibitors against MLL-fusion AMLs and potentially other aggressive tumors with active TLR signaling., (© 2022 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
39. Multi-omics analysis defines highly refractory RAS burdened immature subgroup of infant acute lymphoblastic leukemia.
- Author
-
Isobe T, Takagi M, Sato-Otsubo A, Nishimura A, Nagae G, Yamagishi C, Tamura M, Tanaka Y, Asada S, Takeda R, Tsuchiya A, Wang X, Yoshida K, Nannya Y, Ueno H, Akazawa R, Kato I, Mikami T, Watanabe K, Sekiguchi M, Seki M, Kimura S, Hiwatari M, Kato M, Fukuda S, Tatsuno K, Tsutsumi S, Kanai A, Inaba T, Shiozawa Y, Shiraishi Y, Chiba K, Tanaka H, Kotecha RS, Cruickshank MN, Ishikawa F, Morio T, Eguchi M, Deguchi T, Kiyokawa N, Arakawa Y, Koh K, Aoki Y, Ishihara T, Tomizawa D, Miyamura T, Ishii E, Mizutani S, Wilson NK, Göttgens B, Miyano S, Kitamura T, Goyama S, Yokoyama A, Aburatani H, Ogawa S, and Takita J
- Subjects
- Gene Fusion, Humans, Infant, Transcription Factors genetics, Histone-Lysine N-Methyltransferase genetics, Myeloid-Lymphoid Leukemia Protein genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
KMT2A-rearranged infant acute lymphoblastic leukemia (ALL) represents the most refractory type of childhood leukemia. To uncover the molecular heterogeneity of this disease, we perform RNA sequencing, methylation array analysis, whole exome and targeted deep sequencing on 84 infants with KMT2A-rearranged leukemia. Our multi-omics clustering followed by single-sample and single-cell inference of hematopoietic differentiation establishes five robust integrative clusters (ICs) with different master transcription factors, fusion partners and corresponding stages of B-lymphopoietic and early hemato-endothelial development: IRX-type differentiated (IC1), IRX-type undifferentiated (IC2), HOXA-type MLLT1 (IC3), HOXA-type MLLT3 (IC4), and HOXA-type AFF1 (IC5). Importantly, our deep mutational analysis reveals that the number of RAS pathway mutations predicts prognosis and that the most refractory subgroup of IC2 possesses 100% frequency and the heaviest burden of RAS pathway mutations. Our findings highlight the previously under-appreciated intra- and inter-patient heterogeneity of KMT2A-rearranged infant ALL and provide a rationale for the future development of genomics-guided risk stratification and individualized therapy., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
40. Mechanisms involved in hematopoietic stem cell aging.
- Author
-
Fujino T, Asada S, Goyama S, and Kitamura T
- Subjects
- Epigenesis, Genetic, Hematopoiesis, Cellular Senescence, Hematopoietic Stem Cells physiology
- Abstract
Hematopoietic stem cells (HSCs) undergo progressive functional decline over time due to both internal and external stressors, leading to aging of the hematopoietic system. A comprehensive understanding of the molecular mechanisms underlying HSC aging will be valuable in developing novel therapies for HSC rejuvenation and to prevent the onset of several age-associated diseases and hematological malignancies. This review considers the general causes of HSC aging that range from cell-intrinsic factors to cell-extrinsic factors. In particular, epigenetics and inflammation have been implicated in the linkage of HSC aging, clonality, and oncogenesis. The challenges in clarifying mechanisms of HSC aging have accelerated the development of therapeutic interventions to rejuvenate HSCs, the major goal of aging research; these details are also discussed in this review., (© 2022. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)
- Published
- 2022
- Full Text
- View/download PDF
41. RUNX1 Inhibition Using Lipid Nanoparticle-Mediated Silencing RNA Delivery as an Effective Treatment for Acute Leukemias.
- Author
-
Iida K, Tsuchiya A, Tamura M, Yamamoto K, Kawata S, Ishihara-Sugano M, Kato M, Kitamura T, and Goyama S
- Subjects
- Cell Line, Tumor, Humans, Liposomes, Nanoparticles, RNA, Small Interfering genetics, Core Binding Factor Alpha 2 Subunit genetics, Core Binding Factor Alpha 2 Subunit metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute therapy
- Abstract
Transcription factor RUNX1 plays key roles in the establishment and maintenance of the hematopoietic system. Although RUNX1 has been considered a beneficial tumor suppressor, several recent reports have described the tumor-promoting role of RUNX1 in a variety of hematopoietic neoplasms. In this study, we assessed the effect of RUNX1 depletion in multiple human leukemia cell lines using the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 system, and confirmed that RUNX1 is in fact required for sustaining their leukemic proliferation. To achieve efficient RUNX1 inhibition in leukemia cells, we then examined the effect of lipid nanoparticle (LNP)-mediated delivery of RUNX1-targeting small interfering (si)RNA using two tumor-tropic LNPs. The LNPs containing RUNX1-targeting siRNA were efficiently incorporated into myeloid and T-cell leukemia cell lines and patient-derived primary human acute myeloid leukemia (AML) cells, downregulated RUNX1 expression, induced cell cycle arrest and apoptosis, and exhibited the growth-inhibitory effect in them. In contrast, the LNPs were not efficiently incorporated into normal cord blood CD34
+ cells, indicating their minimum cytotoxicity. Thus, our study highlights RUNX1 as a potential therapeutic target to inhibit leukemogenesis, and provides the LNP-based siRNA delivery as a promising approach to deplete RUNX1 specifically in leukemia cells., Competing Interests: Conflicts of interest disclosure SK and MS work for Toshiba Corporation. S.G. received a research grant from Toshiba Corporation. Other authors declare that they have no conflicts of interest with respect to the contents of this article., (Copyright © 2022 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
42. MDS cells impair osteolineage differentiation of MSCs via extracellular vesicles to suppress normal hematopoiesis.
- Author
-
Hayashi Y, Kawabata KC, Tanaka Y, Uehara Y, Mabuchi Y, Murakami K, Nishiyama A, Kiryu S, Yoshioka Y, Ota Y, Sugiyama T, Mikami K, Tamura M, Fukushima T, Asada S, Takeda R, Kunisaki Y, Fukuyama T, Yokoyama K, Uchida T, Hagihara M, Ohno N, Usuki K, Tojo A, Katayama Y, Goyama S, Arai F, Tamura T, Nagasawa T, Ochiya T, Inoue D, and Kitamura T
- Subjects
- Animals, Hematopoiesis, Hematopoietic Stem Cells metabolism, Mice, Extracellular Vesicles metabolism, Mesenchymal Stem Cells metabolism, Myelodysplastic Syndromes metabolism
- Abstract
Myelodysplastic syndrome (MDS) is a clonal disorder of hematopoietic stem cells (HSCs), characterized by ineffective hematopoiesis and frequent progression to leukemia. It has long remained unresolved how MDS cells, which are less proliferative, inhibit normal hematopoiesis and eventually dominate the bone marrow space. Despite several studies implicating mesenchymal stromal or stem cells (MSCs), a principal component of the HSC niche, in the inhibition of normal hematopoiesis, the molecular mechanisms underlying this process remain unclear. Here, we demonstrate that both human and mouse MDS cells perturb bone metabolism by suppressing the osteolineage differentiation of MSCs, which impairs the ability of MSCs to support normal HSCs. Enforced MSC differentiation rescues the suppressed normal hematopoiesis in both in vivo and in vitro MDS models. Intriguingly, the suppression effect is reversible and mediated by extracellular vesicles (EVs) derived from MDS cells. These findings shed light on the novel MDS EV-MSC axis in ineffective hematopoiesis., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
43. CHIP-associated mutant ASXL1 in blood cells promotes solid tumor progression.
- Author
-
Liu X, Sato N, Shimosato Y, Wang TW, Denda T, Chang YH, Yabushita T, Fujino T, Asada S, Tanaka Y, Fukuyama T, Enomoto Y, Ota Y, Sakamoto T, Kitamura T, and Goyama S
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, Hematopoiesis genetics, Mice, Mutation, Transcription Factors genetics, Tumor Microenvironment, Clonal Hematopoiesis genetics, Neoplasms, Repressor Proteins genetics, Repressor Proteins metabolism
- Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is an age-associated phenomenon characterized by clonal expansion of blood cells harboring somatic mutations in hematopoietic genes, including DNMT3A, TET2, and ASXL1. Clinical evidence suggests that CHIP is highly prevalent and associated with poor prognosis in solid-tumor patients. However, whether blood cells with CHIP mutations play a causal role in promoting the development of solid tumors remained unclear. Using conditional knock-in mice that express CHIP-associated mutant Asxl1 (Asxl1-MT), we showed that expression of Asxl1-MT in T cells, but not in myeloid cells, promoted solid-tumor progression in syngeneic transplantation models. We also demonstrated that Asxl1-MT-expressing blood cells accelerated the development of spontaneous mammary tumors induced by MMTV-PyMT. Intratumor analysis of the mammary tumors revealed the reduced T-cell infiltration at tumor sites and programmed death receptor-1 (PD-1) upregulation in CD8
+ T cells in MMTV-PyMT/Asxl1-MT mice. In addition, we found that Asxl1-MT induced T-cell dysregulation, including aberrant intrathymic T-cell development, decreased CD4/CD8 ratio, and naïve-memory imbalance in peripheral T cells. These results indicate that Asxl1-MT perturbs T-cell development and function, which contributes to creating a protumor microenvironment for solid tumors. Thus, our findings raise the possibility that ASXL1-mutated blood cells exacerbate solid-tumor progression in ASXL1-CHIP carriers., (© 2022 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.)- Published
- 2022
- Full Text
- View/download PDF
44. PRDM paralogs antagonistically balance Wnt/β-catenin activity during craniofacial chondrocyte differentiation.
- Author
-
Shull LC, Lencer ES, Kim HM, Goyama S, Kurokawa M, Costello JC, Jones K, and Artinger KB
- Subjects
- Animals, Cartilage cytology, Cartilage metabolism, Chondrocytes cytology, Chondrocytes metabolism, Chondrogenesis, Chromatin metabolism, Chromatin Assembly and Disassembly, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Gene Expression Regulation, Developmental, MDS1 and EVI1 Complex Locus Protein deficiency, MDS1 and EVI1 Complex Locus Protein genetics, Mice, Mice, Knockout, Neural Crest cytology, Neural Crest metabolism, Regulatory Sequences, Nucleic Acid, Skull cytology, Skull metabolism, Wnt Proteins metabolism, Zebrafish, Zebrafish Proteins deficiency, Zebrafish Proteins genetics, beta Catenin metabolism, Cell Differentiation, MDS1 and EVI1 Complex Locus Protein metabolism, Wnt Signaling Pathway genetics, Zebrafish Proteins metabolism
- Abstract
Cranial neural crest cell (NCC)-derived chondrocyte precursors undergo a dynamic differentiation and maturation process to establish a scaffold for subsequent bone formation, alterations in which contribute to congenital birth defects. Here, we demonstrate that transcription factor and histone methyltransferase proteins Prdm3 and Prdm16 control the differentiation switch of cranial NCCs to craniofacial cartilage. Loss of either paralog results in hypoplastic and disorganized chondrocytes due to impaired cellular orientation and polarity. We show that these proteins regulate cartilage differentiation by controlling the timing of Wnt/β-catenin activity in strikingly different ways: Prdm3 represses whereas Prdm16 activates global gene expression, although both act by regulating Wnt enhanceosome activity and chromatin accessibility. Finally, we show that manipulating Wnt/β-catenin signaling pharmacologically or generating prdm3-/-;prdm16-/- double mutants rescues craniofacial cartilage defects. Our findings reveal upstream regulatory roles for Prdm3 and Prdm16 in cranial NCCs to control Wnt/β-catenin transcriptional activity during chondrocyte differentiation to ensure proper development of the craniofacial skeleton., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
45. Eliminating chronic myeloid leukemia stem cells by IRAK1/4 inhibitors.
- Author
-
Tanaka Y, Takeda R, Fukushima T, Mikami K, Tsuchiya S, Tamura M, Adachi K, Umemoto T, Asada S, Watanabe N, Morishita S, Imai M, Nagata M, Araki M, Takizawa H, Fukuyama T, Lamagna C, Masuda ES, Ito R, Goyama S, Komatsu N, Takaku T, and Kitamura T
- Subjects
- Animals, Antineoplastic Agents pharmacology, Chronic Disease, Drug Resistance, Neoplasm drug effects, Female, Fusion Proteins, bcr-abl metabolism, Humans, Imatinib Mesylate pharmacology, Immune Checkpoint Inhibitors pharmacology, Interleukin-1 Receptor-Associated Kinases metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Male, Mice, Middle Aged, Protein Kinase Inhibitors pharmacology, Interleukin-1 Receptor-Associated Kinases drug effects, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myeloid drug therapy, Neoplastic Stem Cells metabolism
- Abstract
Leukemia stem cells (LSCs) in chronic myeloid leukemia (CML) are quiescent, insensitive to BCR-ABL1 tyrosine kinase inhibitors (TKIs) and responsible for CML relapse. Therefore, eradicating quiescent CML LSCs is a major goal in CML therapy. Here, using a G
0 marker (G0 M), we narrow down CML LSCs as G0 M- and CD27- double positive cells among the conventional CML LSCs. Whole transcriptome analysis reveals NF-κB activation via inflammatory signals in imatinib-insensitive quiescent CML LSCs. Blocking NF-κB signals by inhibitors of interleukin-1 receptor-associated kinase 1/4 (IRAK1/4 inhibitors) together with imatinib eliminates mouse and human CML LSCs. Intriguingly, IRAK1/4 inhibitors attenuate PD-L1 expression on CML LSCs, and blocking PD-L1 together with imatinib also effectively eliminates CML LSCs in the presence of T cell immunity. Thus, IRAK1/4 inhibitors can eliminate CML LSCs through inhibiting NF-κB activity and reducing PD-L1 expression. Collectively, the combination of TKIs and IRAK1/4 inhibitors is an attractive strategy to achieve a radical cure of CML., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
46. CRISPR/Cas9-mediated base-editing enables a chain reaction through sequential repair of sgRNA scaffold mutations.
- Author
-
Fukushima T, Tanaka Y, Adachi K, Masuyama N, Tsuchiya A, Asada S, Ishiguro S, Mori H, Seki M, Yachie N, Goyama S, and Kitamura T
- Subjects
- Cytosine Deaminase genetics, Cytosine Deaminase metabolism, HEK293 Cells, Humans, TATA Box genetics, Thymidine genetics, CRISPR-Cas Systems, DNA Repair, Gene Editing, Mutation, RNA, Guide, CRISPR-Cas Systems genetics
- Abstract
Cell behavior is controlled by complex gene regulatory networks. Although studies have uncovered diverse roles of individual genes, it has been challenging to record or control sequential genetic events in living cells. In this study, we designed two cellular chain reaction systems that enable sequential sgRNA activation in mammalian cells using a nickase Cas9 tethering of a cytosine nucleotide deaminase (nCas9-CDA). In these systems, thymidine (T)-to-cytosine (C) substitutions in the scaffold region of the sgRNA or the TATA box-containing loxP sequence (TATAloxP) are corrected by the nCas9-CDA, leading to activation of the next sgRNA. These reactions can occur multiple times, resulting in cellular chain reactions. As a proof of concept, we established a chain reaction by repairing sgRNA scaffold mutations in 293 T cells. Importantly, the results obtained in yeast or in vitro did not match those obtained in mammalian cells, suggesting that in vivo chain reactions need to be optimized in appropriate cellular contexts. Our system may lay the foundation for building cellular chain reaction systems that have a broad utility in the future biomedical research., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
47. MECOM permits pancreatic acinar cell dedifferentiation avoiding cell death under stress conditions.
- Author
-
Backx E, Wauters E, Baldan J, Van Bulck M, Michiels E, Heremans Y, De Paep DL, Kurokawa M, Goyama S, Bouwens L, Jacquemin P, Houbracken I, and Rooman I
- Subjects
- Animals, Cell Dedifferentiation, Disease Models, Animal, Humans, Mice, Signal Transduction, Acinar Cells metabolism, Cell Death genetics, MDS1 and EVI1 Complex Locus Protein metabolism, Oncogenes genetics
- Abstract
Maintenance of the pancreatic acinar cell phenotype suppresses tumor formation. Hence, repetitive acute or chronic pancreatitis, stress conditions in which the acinar cells dedifferentiate, predispose for cancer formation in the pancreas. Dedifferentiated acinar cells acquire a large panel of duct cell-specific markers. However, it remains unclear to what extent dedifferentiated acini differ from native duct cells and which genes are uniquely regulating acinar cell dedifferentiation. Moreover, most studies have been performed on mice since the availability of human cells is scarce. Here, we applied a non-genetic lineage tracing method of human pancreatic exocrine acinar and duct cells that allowed cell-type-specific gene expression profiling by RNA sequencing. Subsequent to this discovery analysis, one transcription factor that was unique for dedifferentiated acinar cells was functionally characterized. RNA sequencing analysis showed that human dedifferentiated acinar cells expressed genes in "Pathways of cancer" with a prominence of MECOM (EVI-1), a transcription factor that was not expressed by duct cells. During mouse embryonic development, pre-acinar cells also transiently expressed MECOM and in the adult mouse pancreas, MECOM was re-expressed when mice were subjected to acute and chronic pancreatitis, conditions in which acinar cells dedifferentiate. In human cells and in mice, MECOM expression correlated with and was directly regulated by SOX9. Mouse acinar cells that, by genetic manipulation, lose the ability to upregulate MECOM showed impaired cell adhesion, more prominent acinar cell death, and suppressed acinar cell dedifferentiation by limited ERK signaling. In conclusion, we transcriptionally profiled the two major human pancreatic exocrine cell types, acinar and duct cells, during experimental stress conditions. We provide insights that in dedifferentiated acinar cells, cancer pathways are upregulated in which MECOM is a critical regulator that suppresses acinar cell death by permitting cellular dedifferentiation., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
48. A histone modifier, ASXL1, interacts with NONO and is involved in paraspeckle formation in hematopoietic cells.
- Author
-
Yamamoto K, Goyama S, Asada S, Fujino T, Yonezawa T, Sato N, Takeda R, Tsuchiya A, Fukuyama T, Tanaka Y, Yokoyama A, Toya H, Kon A, Nannya Y, Onoguchi-Mizutani R, Nakagawa S, Hirose T, Ogawa S, Akimitsu N, and Kitamura T
- Subjects
- Animals, DNA-Binding Proteins genetics, Female, HL-60 Cells, HeLa Cells, Hematopoiesis, Humans, Mice, Mice, Transgenic, Paraspeckles genetics, RNA-Binding Proteins genetics, Repressor Proteins genetics, THP-1 Cells, DNA-Binding Proteins metabolism, Hematopoietic Stem Cells metabolism, Paraspeckles metabolism, RNA-Binding Proteins metabolism, Repressor Proteins metabolism
- Abstract
Paraspeckles are membraneless organelles formed through liquid-liquid phase separation and consist of multiple proteins and RNAs, including NONO, SFPQ, and NEAT1. The role of paraspeckles and the component NONO in hematopoiesis remains unknown. In this study, we show histone modifier ASXL1 is involved in paraspeckle formation. ASXL1 forms phase-separated droplets, upregulates NEAT1 expression, and increases NONO-NEAT1 interactions through the C-terminal intrinsically disordered region (IDR). In contrast, a pathogenic ASXL mutant (ASXL1-MT) lacking IDR does not support the interaction of paraspeckle components. Furthermore, paraspeckles are disrupted and Nono localization is abnormal in the cytoplasm of hematopoietic stem and progenitor cells (HSPCs) derived from ASXL1-MT knockin mice. Nono depletion and the forced expression of cytoplasmic NONO impair the repopulating potential of HSPCs, as does ASXL1-MT. Our study indicates a link between ASXL1 and paraspeckle components in the maintenance of normal hematopoiesis., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
49. Mutant ASXL1 induces age-related expansion of phenotypic hematopoietic stem cells through activation of Akt/mTOR pathway.
- Author
-
Fujino T, Goyama S, Sugiura Y, Inoue D, Asada S, Yamasaki S, Matsumoto A, Yamaguchi K, Isobe Y, Tsuchiya A, Shikata S, Sato N, Morinaga H, Fukuyama T, Tanaka Y, Fukushima T, Takeda R, Yamamoto K, Honda H, Nishimura EK, Furukawa Y, Shibata T, Abdel-Wahab O, Suematsu M, and Kitamura T
- Subjects
- Aged, Aging metabolism, Aging physiology, Animals, Apoptosis genetics, Cell Cycle genetics, Cell Proliferation genetics, Cells, Cultured, DNA Damage drug effects, DNA Damage genetics, Gene Knock-In Techniques, Hematopoiesis physiology, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells physiology, Humans, Membrane Potential, Mitochondrial drug effects, Membrane Potential, Mitochondrial genetics, Mice, Mice, Transgenic, Mutation, Proto-Oncogene Proteins c-akt metabolism, RNA-Seq, Reactive Oxygen Species pharmacology, Repressor Proteins metabolism, Signal Transduction drug effects, Signal Transduction genetics, Sirolimus pharmacology, Tumor Suppressor Proteins metabolism, Ubiquitin Thiolesterase metabolism, Ubiquitination drug effects, Ubiquitination genetics, Aging genetics, Clonal Hematopoiesis genetics, Hematopoiesis genetics, Hematopoietic Stem Cells metabolism, Repressor Proteins genetics, TOR Serine-Threonine Kinases metabolism
- Abstract
Somatic mutations of ASXL1 are frequently detected in age-related clonal hematopoiesis (CH). However, how ASXL1 mutations drive CH remains elusive. Using knockin (KI) mice expressing a C-terminally truncated form of ASXL1-mutant (ASXL1-MT), we examined the influence of ASXL1-MT on physiological aging in hematopoietic stem cells (HSCs). HSCs expressing ASXL1-MT display competitive disadvantage after transplantation. Nevertheless, in genetic mosaic mouse model, they acquire clonal advantage during aging, recapitulating CH in humans. Mechanistically, ASXL1-MT cooperates with BAP1 to deubiquitinate and activate AKT. Overactive Akt/mTOR signaling induced by ASXL1-MT results in aberrant proliferation and dysfunction of HSCs associated with age-related accumulation of DNA damage. Treatment with an mTOR inhibitor rapamycin ameliorates aberrant expansion of the HSC compartment as well as dysregulated hematopoiesis in aged ASXL1-MT KI mice. Our findings suggest that ASXL1-MT provokes dysfunction of HSCs, whereas it confers clonal advantage on HSCs over time, leading to the development of CH.
- Published
- 2021
- Full Text
- View/download PDF
50. Efficacy of tyrosine kinase inhibitors on a mouse chronic myeloid leukemia model and chronic myeloid leukemia stem cells.
- Author
-
Tanaka Y, Fukushima T, Mikami K, Adachi K, Fukuyama T, Goyama S, and Kitamura T
- Subjects
- Animals, Drug Screening Assays, Antitumor, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl metabolism, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive enzymology, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Mice, Mice, Inbred BALB C, Neoplasms, Experimental enzymology, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Neoplastic Stem Cells pathology, Fusion Proteins, bcr-abl antagonists & inhibitors, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Neoplasms, Experimental drug therapy, Neoplastic Stem Cells enzymology, Protein Kinase Inhibitors pharmacology
- Abstract
Chronic myeloid leukemia (CML) is a hematopoietic stem cell disorder caused by constitutively active BCR-ABL1 tyrosine kinase resulting from the t(9;22) Philadelphia translocation. Imatinib, a BCR-ABL1 tyrosine kinase inhibitor (TKI), is a revolutionary molecular target inhibitor for CML. However, leukemic stem cells (LSCs) eventually become resistant to imatinib and thereby cause relapse. The next-generation BCR-ABL1 TKI dasatinib is also unable to eliminate CML LSCs. On the other hand, the third-generation BCR-ABL1 TKI ponatinib is not well studied in terms of its efficacy on CML LSCs. Here, we evaluate the efficacy of ponatinib against CML LSC-containing lin
- Sca-1+ c-Kit+ (LSK) cells using a mouse CML-like model. To this end, we compared the efficacy of imatinib, dasatinib, and ponatinib on CML LSK cells and showed that ponatinib is more effective at eliminating CML LSK cells. Our results suggest that ponatinib could be potentially useful for achieving treatment-free remission in CML patients., (Copyright © 2020 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.